1
|
Zhang C, Zhang C, Wang K, Wang H. Orchestrating smart therapeutics to achieve optimal treatment in small cell lung cancer: recent progress and future directions. J Transl Med 2023; 21:468. [PMID: 37452395 PMCID: PMC10349514 DOI: 10.1186/s12967-023-04338-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant malignancy with elusive mechanism of pathogenesis and dismal prognosis. Over the past decades, platinum-based chemotherapy has been the backbone treatment for SCLC. However, subsequent chemoresistance after initial effectiveness urges researchers to explore novel therapeutic targets of SCLC. Recent years have witnessed significant improvements in targeted therapy in SCLC. New molecular candidates such as Ataxia telangiectasia and RAD3-related protein (ATR), WEE1, checkpoint kinase 1 (CHK1) and poly-ADP-ribose polymerase (PARP) have shown promising therapeutic utility in SCLC. While immune checkpoint inhibitor (ICI) has emerged as an indispensable treatment modality for SCLC, approaches to boost efficacy and reduce toxicity as well as selection of reliable biomarkers for ICI in SCLC have remained elusive and warrants our further investigation. Given the increasing importance of precision medicine in SCLC, optimal subtyping of SCLC using multi-omics have gradually applied into clinical practice, which may identify more drug targets and better tailor treatment strategies to each individual patient. The present review summarizes recent progress and future directions in SCLC. In addition to the emerging new therapeutics, we also focus on the establishment of predictive model for early detection of SCLC. More importantly, we also propose a multi-dimensional model in the prognosis of SCLC to ultimately attain the goal of accurate treatment of SCLC.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Chenxing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Number 440, Ji Yan Road, Jinan, China.
| |
Collapse
|
2
|
Scalia P, Williams SJ, Fujita-Yamaguchi Y, Giordano A. Cell cycle control by the insulin-like growth factor signal: at the crossroad between cell growth and mitotic regulation. Cell Cycle 2023; 22:1-37. [PMID: 36005738 PMCID: PMC9769454 DOI: 10.1080/15384101.2022.2108117] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In proliferating cells and tissues a number of checkpoints (G1/S and G2/M) preceding cell division (M-phase) require the signal provided by growth factors present in serum. IGFs (I and II) have been demonstrated to constitute key intrinsic components of the peptidic active fraction of mammalian serum. In vivo genetic ablation studies have shown that the cellular signal triggered by the IGFs through their cellular receptors represents a non-replaceable requirement for cell growth and cell cycle progression. Retroactive and current evaluation of published literature sheds light on the intracellular circuitry activated by these factors providing us with a better picture of the pleiotropic mechanistic actions by which IGFs regulate both cell size and mitogenesis under developmental growth as well as in malignant proliferation. The present work aims to summarize the cumulative knowledge learned from the IGF ligands/receptors and their intracellular signaling transducers towards control of cell size and cell-cycle with particular focus to their actionable circuits in human cancer. Furthermore, we bring novel perspectives on key functional discriminants of the IGF growth-mitogenic pathway allowing re-evaluation on some of its signal components based upon established evidences.
Collapse
Affiliation(s)
- Pierluigi Scalia
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states,CONTACT Pierluigi Scalia ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA9102, USA
| | - Stephen J Williams
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states
| | - Yoko Fujita-Yamaguchi
- Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Antonio Giordano
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,School of Medical Biotechnology, University of Siena, Italy
| |
Collapse
|
3
|
Zhang C, Wang H. Accurate treatment of small cell lung cancer: Current progress, new challenges and expectations. Biochim Biophys Acta Rev Cancer 2022; 1877:188798. [PMID: 36096336 DOI: 10.1016/j.bbcan.2022.188798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/19/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022]
Abstract
Small cell lung cancer (SCLC) is a deadly disease with poor prognosis. Fast growing speed, inclination to metastasis, enrichment in cancer stem cells altogether constitute its aggressive nature. In stark contrast to non-small cell lung cancer (NSCLC) that strides vigorously on the road to precision oncology, SCLC has been on the embryonic path to achieve effective personalized treatments. The survival of patients with SCLC have not been improved greatly, which could be possibly due to our inadequate understanding of genetic alterations of SCLC. Recently, encouraging effects have been observed in patients with SCLC undergoing immunotherapy. However, exciting results have only been observed in a small fraction of patients with SCLC, warranting biomarkers predictive of responses as well as novel therapeutic strategies. In addition, SCLC has previously been viewed to be homogeneous. However, perspectives have been changed thanks to the advances in sequencing techniques and platforms, which unfolds the complex heterogeneity of SCLC both genetically and non-genetically, rendering the treatment of SCLC a further step forward into the precision era. To outline the road of SCLC towards precision oncology, we summarize the progresses and achievements made in precision treatment in SCLC in genomic, transcriptomic, epigenetic, proteomic and metabolic dimensions. Moreover, we conclude relevant therapeutic vulnerabilities in SCLC. Clinically tested drugs and clinical trials have also been demonstrated. Ultimately, we look into the opportunities and challenges ahead to advance the individualized treatment in pursuit of improved survival for patients with SCLC.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
4
|
Costa-Silva DR, Barros-Oliveira MDC, Alves-Ribeiro FA, Campos-Verdes LC, Nery Junior EDJ, Vieira-Valença SF, de Vasconcelos-Valença RJ, Soares VM, Pinho-Sobral AL, Sousa EB, Lopes-Costa PV, dos Santos AR, Viana JL, Cardoso AC, Luz-Borges VM, Pereira RDO, Tavares CB, Silva VC, Rodrigues-Junior DM, Gebrim LH, da Silva BB. Assessment of IGF-1 expression in the peripheral blood of women with recurrent breast cancer. Medicine (Baltimore) 2020; 99:e22890. [PMID: 33120836 PMCID: PMC7581150 DOI: 10.1097/md.0000000000022890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Breast cancer is the most common malignancy affecting women worldwide. The insulin-like growth factor 1 (IGF-1) gene encodes a protein responsible for a wide variety of physiological processes, including differentiation and cell proliferation. Despite several studies on tumor tissues, no study has evaluated IGF-1 expression in the peripheral blood of women with recurrent breast cancer.In this cross-sectional study, IGF-1 expression in the peripheral blood of 146 women with breast cancer treated approximately 5 years ago was quantified by quantitative reverse transcription polymerase chain. The women were divided into 2 groups: non-recurrence (n = 85) and recurrence (n = 61). Statistical analysis of the data was performed using ANOVA, Mann-Whitney, and Chi-squared tests (P < .05).The results showed no significant difference in IGF-1 expression between the non-recurrence and recurrence groups (P = .988). In the subgroups of patients with lymph node involvement, no statistically significant difference was observed in IGF-1 expression between women with recurrence and those non-recurrence (P = .113). In patients without lymph node metastases, IGF-1 messenger ribonucleic acid (mRNA) expression levels were significantly higher in the non-recurrence group than in the recurrence group (P = .019). Furthermore, using the median IGF-1 mRNA expression as the cutoff point, it was obtained a statistically significant difference in tumor histological grade among women with recurrent breast cancer (P = .042).These data showed significantly higher IGF-1 expression in women without lymph node metastases in the non-recurrence group compared with the recurrence group. In addition, a significant difference was observed in median IGF-1 mRNA expression in relation to tumor histological grade in women with recurrent breast cancer.
Collapse
Affiliation(s)
- Danylo Rafhael Costa-Silva
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | | | | | | | - Elmo de Jesus Nery Junior
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | | | | | - Veronica Mendes Soares
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | | | | | | | | | | | | | | | - Renato de Oliveira Pereira
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | - Cleciton Braga Tavares
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | - Vladimir Costa Silva
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
| | | | - Luiz Henrique Gebrim
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
- Department of Mastology, Perola Byington Hospital, São Paulo, Brazil
| | - Benedito Borges da Silva
- Department of Health, Northeast, Postgraduate Program of the Northeast Network of Biotechnology (RENORBIO)
- Getulio Vargas Hospital, Federal University of Piaui, Teresina, Piaui
| |
Collapse
|
5
|
Mulshine JL, Ujhazy P, Antman M, Burgess CM, Kuzmin I, Bunn PA, Johnson BE, Roth JA, Pass HI, Ross SM, Aldige CR, Wistuba II, Minna JD. From clinical specimens to human cancer preclinical models-a journey the NCI-cell line database-25 years later. J Cell Biochem 2020; 121:3986-3999. [PMID: 31803961 PMCID: PMC7496084 DOI: 10.1002/jcb.29564] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 11/13/2019] [Indexed: 01/24/2023]
Abstract
The intramural the National Cancer Institute (NCI) and more recently the University of Texas Southwestern Medical Center with many different collaborators comprised a complex, multi-disciplinary team that collaborated to generated large, comprehensively annotated, cell-line related research resources which includes associated clinical, and molecular characterization data. This material has been shared in an anonymized fashion to accelerate progress in overcoming lung cancer, the leading cause of cancer death across the world. However, this cell line collection also includes a range of other cancers derived from patient-donated specimens that have been remarkably valuable for other types of cancer and disease research. A comprehensive analysis conducted by the NCI Center for Research Strategy of the 278 cell lines reported in the original Journal of Cellular Biochemistry Supplement, documents that these cell lines and related products have since been used in more than 14 000 grants, and 33 207 published scientific reports. This has resulted in over 1.2 million citations using at least one cell line. Many publications involve the use of more than one cell line, to understand the value of the resource collectively rather than individually; this method has resulted in 2.9 million citations. In addition, these cell lines have been linked to 422 clinical trials and cited by 4700 patents through publications. For lung cancer alone, the cell lines have been used in the research cited in the development of over 70 National Comprehensive Cancer Network clinical guidelines. Finally, it must be underscored again, that patient altruism enabled the availability of this invaluable research resource.
Collapse
Affiliation(s)
- James L. Mulshine
- Center for Healthy Aging, Department of Internal MedicineRush UniversityChicagoIllinois
| | - Peter Ujhazy
- Translational Research Program, Division of Cancer Treatment and DiagnosisNational Cancer InstituteRockvilleMaryland
| | - Melissa Antman
- Center for Research StrategyNational Cancer InstituteBethesdaMaryland
| | | | - Igor Kuzmin
- Translational Research Program, Division of Cancer Treatment and DiagnosisNational Cancer InstituteRockvilleMaryland
| | - Paul A. Bunn
- University of Colorado Cancer CenterUniversity of Colorado Cancer CenterAuroraColorado
| | - Bruce E. Johnson
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusetts
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, Division of SurgeryThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Harvey I. Pass
- Department of Cardiothoracic SurgeryNew York University Langone Medical CenterNew YorkNew York
| | - Sheila M. Ross
- AdvocacyLung Cancer AllianceAnnapolisMaryland,MemberIASLC Early Detection and Screening CommitteeAuroraColorado
| | | | - Ignacio I. Wistuba
- Department of Translational Molecular PathologyUT MD Anderson Cancer CenterHoustonTexas
| | - John D Minna
- Nancy B. and Jake L. Hamon Center for Therapeutic Oncology ResearchUT Southwestern Medical CenterDallasTexas
| |
Collapse
|
6
|
Kern JA, Kim J, Foster DG, Mishra R, Gardner EE, Poirier JT, Rivard C, Yu H, Finigan JH, Dowlati A, Rudin CM, Tan AC. Role of mTOR As an Essential Kinase in SCLC. J Thorac Oncol 2020; 15:1522-1534. [PMID: 32599072 DOI: 10.1016/j.jtho.2020.05.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/17/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES SCLC represents 15% of all lung cancer diagnoses in the United States and has a particularly poor prognosis. We hypothesized that kinases regulating SCLC survival pathways represent therapeutically targetable vulnerabilities whose inhibition may improve SCLC outcome. METHODS A short-hairpin RNA (shRNA) library targeting all human kinases was introduced in seven chemonaive patient-derived xenografts (PDX) and the cells were cultured in vitro and in vivo. On harvest, lost or depleted shRNAs were considered as regulating-cell survival pathways and deemed essential kinases. RESULTS Unsupervised hierarchical cluster analysis of recovered shRNAs separated the PDXs into two clusters, suggesting kinase-based heterogeneity among the SCLC PDXs. A total of 23 kinases were identified as essential in two or more PDXs, with mechanistic Target of Rapamycin (mTOR) a candidate essential kinase in four. mTOR phosphorylation status correlated with PDX sensitivity to mTOR kinase inhibition, and mTOR inhibition sensitized the PDX to cisplatin and etoposide. In the PDX in which mTOR was defined as essential, mTOR inhibition caused a 43% decrease in tumor volume at 21 days (p < 0.01). Combining mTOR inhibition with cisplatin and etoposide decreased PDX tumor volume 96% compared with cisplatin and etoposide alone at 70 days (p < 0.002). Chemoresistance did not develop with the combination of mTOR inhibition and cisplatin and etoposide in mTOR-essential PDX over 105 days. The prevalence of phospho-mTOR-Ser-2448 in a tissue microarray of chemonaive SCLC was 27%, thus, identifying an important SCLC subtype that might benefit from the addition of mTOR inhibition to standard chemotherapy. CONCLUSIONS These studies reveal that kinases can define SCLC subgroups, can identify therapeutic vulnerabilities, and can potentially be used to optimize therapeutic approaches. Significance We used functional genomics to identify kinases regulating SCLC survival. mTOR was identified as essential in a subset of PDXs. mTOR inhibition decreased PDX growth, sensitized PDX to cisplatin and etoposide, and prevented chemoresistance.
Collapse
Affiliation(s)
- Jeffrey A Kern
- Department of Medicine, Oncology Division, National Jewish Health, Denver, Colorado.
| | - Jihye Kim
- Department of Medicine, University of Colorado, Denver, Colorado
| | - Daniel G Foster
- Department of Medicine, Oncology Division, National Jewish Health, Denver, Colorado
| | - Rangnath Mishra
- Department of Medicine, Oncology Division, National Jewish Health, Denver, Colorado
| | - Eric E Gardner
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - John T Poirier
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | | | - Hui Yu
- Department of Medicine, University of Colorado, Denver, Colorado
| | - James H Finigan
- Department of Medicine, Oncology Division, National Jewish Health, Denver, Colorado
| | - Afshin Dowlati
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | - Aik-Choon Tan
- Department of Medicine, University of Colorado, Denver, Colorado; Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
7
|
Schramm HM. The Epithelial-Myeloid-Transition (EMyeT) of cancer cells as a wrongly perceived primary inflammatory process eventually progressing to a bone remodeling malignancy: the alternative pathway for Epithelial- Mesenchymal-Transition hypothesis (EMT)? J Cancer 2019; 10:3798-3809. [PMID: 31333797 PMCID: PMC6636288 DOI: 10.7150/jca.31364] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer cells express multiple markers expressed by mesenchymal as well as myeloid cells in common and in addition specific markers of the myeloid lineages, especially those of dendritic cells, macrophages and preosteoclasts. It has also been possible to identify monocyte-macrophage gene clusters in cancer cell specimens as well as in cancer cell lines. Accordingly, like myeloid cells cancer cells often express pro-inflammatory cytokines, and consequently the carcinoma may be perceived by the organism as a primary inflammatory process comparable to the immune inflammatory reactions in the eye or in the case of arthritis. This would explain why a carcinoma may induce a certain alarm state in the organism by increasing a fatal sympathetic tone in the patient, supplying the carcinomas with nutrients at the cost of other requirements, inducing tolerance against the cancer cells mistaken as myeloid cells, provoking fibrosis and neoangiogenesis, and increasing inflammatory cells at the carcinoma site. This seemingly inflammatory process of Epithelial-Myeloid-Transition (EMyeT) is superimposed by the progression of part of the myeloid cancer cells to stages comparable to preosteoclasts and osteoclasts, and their development to metastasizing carcinomas often at the site of bone. This concept of carcinogenesis and malignant progression described here challenges the widely accepted EMT-hypotheses and could deliver the rationale for the various peculiar aspects of cancer and the variety of therapeutic antitumoral measures.
Collapse
Affiliation(s)
- Henning M Schramm
- Institute for Integral Cancer Research (IFIK), CH-4144 Arlesheim/Switzerland
| |
Collapse
|
8
|
Franks SE, Briah R, Jones RA, Moorehead RA. Unique roles of Akt1 and Akt2 in IGF-IR mediated lung tumorigenesis. Oncotarget 2016; 7:3297-316. [PMID: 26654940 PMCID: PMC4823107 DOI: 10.18632/oncotarget.6489] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 11/16/2015] [Indexed: 01/22/2023] Open
Abstract
AKT is a serine-threonine kinase that becomes hyperactivated in a number of cancers including lung cancer. Based on AKT's association with malignancy, molecules targeting AKT have entered clinical trials for solid tumors including lung cancer. However, the AKT inhibitors being evaluated in clinical trials indiscriminately inhibit all three AKT isoforms (AKT1-3) and it remains unclear whether AKT isoforms have overlapping or divergent functions. Using a transgenic mouse model where IGF-IR overexpression drives lung tumorigenesis, we found that loss of Akt1 inhibited while loss of Akt2 enhanced lung tumor development. Lung tumors that developed in the absence of Akt2 were less likely to appear as discrete nodules and more frequently displayed a dispersed growth pattern. RNA sequencing revealed a number of genes differentially expressed in lung tumors lacking Akt2 and five of these genes, Actc1, Bpifa1, Mmp2, Ntrk2, and Scgb3a2 have been implicated in human lung cancer. Using 2 human lung cancer cell lines, we observed that a selective AKT1 inhibitor, A-674563, was a more potent regulator of cell survival than the pan-AKT inhibitor, MK-2206. This study suggests that compounds selectively targeting AKT1 may prove more effective than compounds that inhibit all three AKT isoforms at least in the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- S Elizabeth Franks
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Ritesh Briah
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Robert A Jones
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Roger A Moorehead
- Department of Biomedical Science, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
9
|
Papaioannou A, Kuyucak S, Kuncic Z. Elucidating the Activation Mechanism of the Insulin-Family Proteins with Molecular Dynamics Simulations. PLoS One 2016; 11:e0161459. [PMID: 27548502 PMCID: PMC4993506 DOI: 10.1371/journal.pone.0161459] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 08/05/2016] [Indexed: 12/27/2022] Open
Abstract
The insulin-family proteins bind to their own receptors, but insulin-like growth factor II (IGF-II) can also bind to the A isoform of the insulin receptor (IR-A), activating unique and alternative signaling pathways from those of insulin. Although extensive studies of insulin have revealed that its activation is associated with the opening of the B chain-C terminal (BC-CT), the activation mechanism of the insulin-like growth factors (IGFs) still remains unknown. Here, we present the first comprehensive study of the insulin-family proteins comparing their activation process and mechanism using molecular dynamics simulations to reveal new insights into their specificity to the insulin receptor. We have found that all the proteins appear to exhibit similar stochastic dynamics in their conformational change to an active state. For the IGFs, our simulations show that activation involves two opening locations: the opening of the BC-CT section away from the core, similar to insulin; and the additional opening of the BC-CT section away from the C domain. Furthermore, we have found that these two openings occur simultaneously in IGF-I, but not in IGF-II, where they can occur independently. This suggests that the BC-CT section and the C domain behave as a unified domain in IGF-I, but as two independent domains in IGF-II during the activation process, implying that the IGFs undergo different activation mechanisms for receptor binding. The probabilities of the active and inactive states of the proteins suggest that IGF-II is hyperactive compared to IGF-I. The hinge residue and the hydrophobic interactions in the core are found to play a critical role in the stability and activity of IGFs. Overall, our simulations have elucidated the crucial differences and similarities in the activation mechanisms of the insulin-family proteins, providing new insights into the molecular mechanisms responsible for the observed differences between IGF-I and IGF-II in receptor binding.
Collapse
Affiliation(s)
- Anastasios Papaioannou
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Physics, University of Sydney, Sydney, NSW, Australia
- * E-mail: (AP); (ZK)
| | - Serdar Kuyucak
- School of Physics, University of Sydney, Sydney, NSW, Australia
| | - Zdenka Kuncic
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Physics, University of Sydney, Sydney, NSW, Australia
- * E-mail: (AP); (ZK)
| |
Collapse
|
10
|
Daikuhara S, Uehara T, Higuchi K, Hosaka N, Iwaya M, Maruyama Y, Matsuda K, Arakura N, Tanaka E, Ota H. Insulin-Like Growth Factor II mRNA-Binding Protein 3 (IMP3) as a Useful Immunohistochemical Marker for the Diagnosis of Adenocarcinoma of Small Intestine. Acta Histochem Cytochem 2015; 48:193-204. [PMID: 26855452 PMCID: PMC4731852 DOI: 10.1267/ahc.15021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/26/2015] [Indexed: 11/29/2022] Open
Abstract
The biological characteristics and roles of insulin-like growth factor II mRNA-binding protein 3 protein (IMP3) expression in small-intestinal adenocarcinoma were investigated. The value of IMP3 immunostaining in the diagnosis of small-intestinal epithelial lesions was also evaluated. Immunohistochemical expression of IMP3 in normal small-intestinal mucosa adjacent to adenoma and adenocarcinoma lesions, and inflamed duodenal and ileal mucosa was analyzed. Samples assessed were: duodenal ulcer (n=6), Crohn’s disease (n=5), low-grade small-intestinal adenoma (n=10), high-grade small-intestinal adenoma (n=13), small-intestinal adenocarcinoma (n=23), lymph node metastases (LNM; n=7), and preoperative biopsies of small-intestinal adenocarcinoma (n=6). Immunohistochemical expression of Ki-67 and p53 was also analyzed in adenoma and adenocarcinoma samples. IMP3 was not expressed in normal epithelium, but weakly expressed in reparative epithelium. Meanwhile, increased IMP3 expression was associated with a higher degree of dysplasia in adenomas, higher T classification, LNM, Ki-67 positivity, histological differentiation, and lower 5-year disease-free survival, but not p53 expression in adenocarcinoma. IMP3 expression appears to be a late event in the small-intestinal carcinogenesis. Assessing the IMP3 staining pattern can be useful in the diagnosis of small-intestinal epithelial lesions when used in conjunction with other histological criteria.
Collapse
Affiliation(s)
- Seiichi Daikuhara
- Department of Gastroenterology, Shinshu University School of Medicine
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine
| | | | - Noriko Hosaka
- Department of Laboratory Medicine, Nagano Municipal Hospital
| | - Mai Iwaya
- Department of Pathology, Nagano Municipal Hospital
| | - Yasuhiro Maruyama
- Department of Gastroenterology, Shinshu University School of Medicine
| | | | | | - Eiji Tanaka
- Department of Gastroenterology, Shinshu University School of Medicine
| | - Hiroyoshi Ota
- Department of Biomedical Laboratory Sciences, School of Health Sciences, Shinshu University School of Medicine
| |
Collapse
|
11
|
Bernstein AM, Golubic M, Roizen MF. The impact of protein on chronic disease risk should be considered in studies of weight loss. Am J Clin Nutr 2015; 101:1097-8. [PMID: 25934865 DOI: 10.3945/ajcn.114.105189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Adam M Bernstein
- From the Wellness Institute, Cleveland Clinic, Lyndhurst, OH (e-mail: )
| | - Mladen Golubic
- From the Wellness Institute, Cleveland Clinic, Lyndhurst, OH (e-mail: )
| | - Michael F Roizen
- From the Wellness Institute, Cleveland Clinic, Lyndhurst, OH (e-mail: )
| |
Collapse
|
12
|
NCIC CTG IND.190 phase I trial of dalotuzumab (MK-0646) in combination with cisplatin and etoposide in extensive-stage small-cell lung cancer. J Thorac Oncol 2014; 9:410-3. [PMID: 24518092 DOI: 10.1097/jto.0000000000000058] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The insulin-like growth factor receptor is a potential target in small-cell lung cancer. We conducted a phase I study of cisplatin, etoposide plus dalotuzumab. Two dose levels of dalotuzumab (DL1 5 mg/kg, DL2 10mg/kg IV weekly) were evaluated in combination with cisplatin (25 mg/m²) and etoposide (100 mg/m²) IV D1-3, every 21 days, for patients with chemotherapy-naive extensive-stage small-cell lung cancer. Primary outcome was determination of the recommended phase 2 dose. Secondary outcomes included response rate and toxicity. Twelve patients were treated (DL1, 3 and DL2, 9). The median age was 63 years (48-70), with six males and six females. The majority of patients were Eastern Cooperative Oncology Group 1 and had four or more sites of disease. No dose-limiting toxicities were observed in DL1 or DL2, although one patient died from neutropenic sepsis in an expanded cohort at DL2. The recommended phase 2 dose of dalotuzumab was 10 mg/kg/week. The confirmed objective response rate was 67% (partial response 8, stable disease 2, progressive disease 1, nonevaluable 1). Grade 3 or higher toxicities (any cycle) occurring in more than one patient included: neutropenia (92%); thrombocytopenia (25%); leukopenia (50%); anemia (17%); fatigue (33%); joint pain (17%); thrombosis (25%). Grade 2 or 3 hyperglycemia was observed in one of three (DL1) and five of nine (DL2) patients. Eight serious adverse events (thrombosis, febrile neutropenia, infection, syncope, fatigue [2], dyspnea, back pain) were observed in three patients. Dalotuzumab can be combined at full dose with standard doses of cisplatin and etoposide. The observed toxicities are consistent with that expected from cisplatin and etoposide except for hyperglycemia, which seems to be dose dependent.
Collapse
|
13
|
Park SJ, Ryu J, Kim IH, Choi YH, Nam TJ. Induction of apoptosis by a peptide from Porphyra yezoensis: regulation of the insulin-like growth factor I receptor signaling pathway in MCF-7 cells. Int J Oncol 2014; 45:1011-6. [PMID: 24970277 PMCID: PMC4121416 DOI: 10.3892/ijo.2014.2509] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/16/2014] [Indexed: 11/24/2022] Open
Abstract
This study examined how PPY, a peptide from Porphyra yezoensis, regulates multiple cell growth-related signaling pathways in MCF-7 cells. This study determined that PPY induces cell cycle arrest and inhibits the IGF-IR signaling pathway. Cell proliferation studies revealed that PPY induced cell death in a dose-dependent manner. Expression levels of IGF-IR were decreased in MCF-7 cells by PPY in a dose-dependent manner. These results indicate that inhibition of the IGF-IR pathway is also involved in PPY induced proliferation of MCF-7 cells. In addition, these data demonstrated that PPY induces cell cycle arrest and activates apoptosis.
Collapse
Affiliation(s)
- Su-Jin Park
- Department of Food and Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - Jina Ryu
- Department of Food and Science, Pukyong National University, Busan 608-737, Republic of Korea
| | - In-Hye Kim
- Institute of Fisheries Science, Pukyong National University, Busan 619-911, Republic of Korea
| | - Youn-Hee Choi
- Institute of Fisheries Science, Pukyong National University, Busan 619-911, Republic of Korea
| | - Taek-Jeong Nam
- Department of Food and Science, Pukyong National University, Busan 608-737, Republic of Korea
| |
Collapse
|
14
|
Aljada A, Saleh AM, Al Suwaidan S. Modulation of insulin/IGFs pathways by sirtuin-7 inhibition in drug-induced chemoreistance. Diagn Pathol 2014; 9:94. [PMID: 24885964 PMCID: PMC4229859 DOI: 10.1186/1746-1596-9-94] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 04/15/2014] [Indexed: 01/08/2023] Open
Abstract
Background Insulin and insulin-like growth factors (IGFs) are key regulators of metabolism and growth. Recent evidences suggest a key role of these pathways in non-classical tissues and the metabolic pathways by which these hormones exert their effects in neoplasia is unclear. Aims To study insulin/IGFs pathways in drug sensitive and resistant cancer cells representing breast cancer (MCF-7), osteosarcoma (SaOS-2), and ovarian cancer (A2780) and to examine the effect of Sirtuin-7 (Sirt7) inhibition on insulin/IGFs pathways in MCF-7 cell line. Methods Drug resistant cells were generated by continuous incubation of parental cell lines with stepwise increases in Doxorubicin or Cisplatin over a period of 3 to 6 months. MCF-7 cells were transfected with cloned hairpin siRNA template for Sirt7 using the Amaxa GmbH transfection system. mRNA expression of Sirt7, INSR, IRS-1, IRS-2, IRS-4, IGF-1, IGF-2, MDR-1, MRP-1, BCRP was measured by qPCR and Sirt7 by standard Western blotting. FITC-insulin uptake was imaged with Leica Confocal Microscope. Results Insulin receptor (INSR), insulin receptor substrate-1 (IRS-1) were inhibited in drug-induced resistance, whereas IRS-2 was significantly induced in all the chemoresistant cells tested when compared to their parental counterparts. IGF-1 and IGF-2 were also upregulated in all the drug resistant cells tested. Sirt7 was significantly reduced in all chemoresistant cells tested. Knockdown of Sirt7 expression in human breast MCF-7 cell line by siRNA induced premature senescence-like phenotype and multi-drug resistance, suggesting that this gene may play an active role in regulating cancer cell response to stress. Suppression of Sirt7 selectively inhibited INSR and IRS-1, whereas it had minimal effect on that of IRS-2. Sirt7 suppression in MCF-7 also inhibited insulin uptake. Additionally, Sirt7 inhibition upregulated IGF-1, IGF-2 and IGFR expression. Conclusion Our data demonstrate that stress-induced Sirt7 inhibition significantly increases stress resistance and modulates insulin/IGF-1 signaling pathways. More importantly, this study links Sir2 family proteins to insulin/IGF signaling in drug-induced stress resistance in neoplasia. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1135426681234493
Collapse
Affiliation(s)
- Ahmad Aljada
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, P, O, Box 22490, Riyadh 11426, Kingdom of Saudi Arabia.
| | | | | |
Collapse
|
15
|
Jiang L, Zhu W, Streicher K, Morehouse C, Brohawn P, Ge X, Dong Z, Yin X, Zhu G, Gu Y, Ranade K, Higgs BW, Yao Y, Huang J. Increased IR-A/IR-B ratio in non-small cell lung cancers associates with lower epithelial-mesenchymal transition signature and longer survival in squamous cell lung carcinoma. BMC Cancer 2014; 14:131. [PMID: 24571613 PMCID: PMC3974050 DOI: 10.1186/1471-2407-14-131] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 02/12/2014] [Indexed: 02/06/2023] Open
Abstract
Background To evaluate the insulin receptor isoform mRNA expression status in non-small cell lung cancer (NSCLC) patients. Methods RNA-seq data from 614 NSCLC [355 adenocarcinomas (LUAD) and 259 squamous cell carcinomas (LUSC)] and 92 normal lung specimens were obtained from The Cancer Genome Atlas (TCGA) to evaluate the mRNA expression of insulin receptor isoform A (IR-A) and insulin receptor isoform B (IR-B). The differential expression status of the insulin receptor isoforms in NSCLC patients was confirmed using qRT-PCR assays with lung cancer cDNA arrays and primary tumor samples. Results The mRNA expression levels of IR-B were significantly lower in some NSCLC samples compared to normal lung specimens, including both LUAD and LUSC. Notably, no IR-B transcripts were detected - only the IR-A isoform was expressed in 11% of NSCLC patients. This decrease in IR-B expression contributed to an elevated IR-A/IR-B ratio, which was also associated with lower epithelial-mesenchymal transition gene signatures in NSCLC and longer patient survival under standard of care in LUSC. In addition to NSCLC, RNA-seq data from TCGA revealed a similar increase in IR-A/IR-B ratio in many other cancer types, with high prevalence in acute myeloid leukemia, glioblastoma multiforme, and brain lower grade glioma. Conclusions Our results indicate a common reduction of the mRNA expression level of IR-B and an increased IR-A/IR-B mRNA ratio in NSCLC and other tumor types. The relationship of altered IR-A/IR-B ratios with cancer progression and patient survival should be prospectively explored in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yihong Yao
- MedImmune Inc,, LLC, One MedImmune Way, 20878 Gaithersburg, MD, USA.
| | | |
Collapse
|
16
|
The effect of IGF-I receptor blockade for human esophageal squamous cell carcinoma and adenocarcinoma. Tumour Biol 2013; 35:973-85. [PMID: 24026884 DOI: 10.1007/s13277-013-1131-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/21/2013] [Indexed: 12/31/2022] Open
Abstract
Insulin-like growth factor-I receptor (IGF-IR) signaling is required for carcinogenicity and tumor development, and this pathway has not been well studied in human esophageal carcinomas. Esophageal cancer is one of the human cancers with the worst prognosis and has two main histologies: squamous cell carcinomas (ESCC) and adenocarcinoma (EAC). Previously, we have reported that detection of the IGF axis may be useful for the prediction of recurrence and poor prognosis of ESCC. We have also shown the successful therapy for several gastrointestinal cancers using recombinant adenoviruses expressing dominant negative IGF-IR (ad-IGF-IR/dn). The aim of this study is to develop potential targeted therapeutics to IGF-IR and to assess the effect of IGF-IR blockade in both of these types of esophageal cancer. We determined immunohistochemical expression of IGF-IR in a tissue microarray. We then assessed the effect of IGF-IR blockade on signal transduction, proliferation, apoptosis, and motility. Ad-IGF-IR/dn, a tyrosine kinase inhibitor, BMS-536924, and adenovirus expressing shRNA for IGF-IR were used. IGF-IR expression was common in both tumor types but not in normal tissues. IGF-IR was detected in metastatic sites at similar levels compared to the primary site. IGF-IR inhibition suppressed proliferation and colony formation in both cancers. IGF-IR blockades up-regulated both stress- and chemotherapy-induced apoptosis and reduced migration. Although IGF-IR/dn blocked ligand-induced activation of Akt-1 mainly, BMS-536924 effectively blocked both activation of Akt and MAPK. The IGF axis might play a key role in tumor progression of esophageal carcinomas. The IGF-IR targeting strategies might thus be useful anticancer therapeutics for human esophageal malignancies.
Collapse
|
17
|
Vincent EE, Elder DJE, Curwen J, Kilgour E, Hers I, Tavaré JM. Targeting non-small cell lung cancer cells by dual inhibition of the insulin receptor and the insulin-like growth factor-1 receptor. PLoS One 2013; 8:e66963. [PMID: 23826179 PMCID: PMC3691253 DOI: 10.1371/journal.pone.0066963] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/14/2013] [Indexed: 11/18/2022] Open
Abstract
Phase III trials of the anti-insulin-like growth factor-1 receptor (IGF1R) antibody figitumumab in non-small cell lung cancer (NSCLC) patients have been discontinued owing to lack of survival benefit. We investigated whether inhibition of the highly homologous insulin receptor (IR) in addition to the IGF1R would be more effective than inhibition of the IGF1R alone at preventing the proliferation of NSCLC cells. Signalling through IGF1R and IR in the NSCLC cell lines A549 and Hcc193 was stimulated by a combination of IGF1, IGF2 and insulin. It was inhibited by antibodies that block ligand binding, αIR3 (IGF1R) and IR47-9 (IR), and by the ATP-competitive small molecule tyrosine kinase inhibitors AZ12253801 and NVPAWD742 which inhibit both IGF1R and IR tyrosine kinases. The effect of inhibitors was determined by an anchorage-independent proliferation assay and by analysis of Akt phosphorylation. In Hcc193 cells the reduction in cell proliferation and Akt phosphorylation due to anti-IGF1R antibody was enhanced by antibody-mediated inhibition of the IR whereas in A549 cells, with a relatively low IR:IGF1R expression ratio, it was not. In each cell line proliferation and Akt phosphorylation were more effectively inhibited by AZ12253801 and NVPAWD742 than by combined αIR3 and IR47-9. When the IGF1R alone is inhibited, unencumbered signalling through the IR can contribute to continued NSCLC cell proliferation. We conclude that small molecule inhibitors targeting both the IR and IGF1R more effectively reduce NSCLC cell proliferation in a manner independent of the IR:IGF1R expression ratio, providing a therapeutic rationale for the treatment of this disease.
Collapse
Affiliation(s)
- Emma E. Vincent
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Douglas J. E. Elder
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jon Curwen
- CIRA Discovery, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Elaine Kilgour
- CIRA Discovery, AstraZeneca, Alderley Park, Macclesfield, United Kingdom
| | - Ingeborg Hers
- School of Physiology and Pharmacology, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Jeremy M. Tavaré
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
18
|
Zinn RL, Gardner EE, Marchionni L, Murphy SC, Dobromilskaya I, Hann CL, Rudin CM. ERK phosphorylation is predictive of resistance to IGF-1R inhibition in small cell lung cancer. Mol Cancer Ther 2013; 12:1131-9. [PMID: 23515613 DOI: 10.1158/1535-7163.mct-12-0618] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
New therapies are critically needed to improve the outcome for patients with small cell lung cancer (SCLC). Insulin-like growth factor 1 receptor (IGF-1R) inhibition is a potential treatment strategy for SCLC: the IGF-1R pathway is commonly upregulated in SCLC and has been associated with inhibition of apoptosis and stimulation of proliferation through downstream signaling pathways, including phosphatidylinositol-3-kinase-Akt and mitogen-activated protein kinase. To evaluate potential determinants of response to IGF-1R inhibition, we assessed the relative sensitivity of 19 SCLC cell lines to OSI-906, a small molecule inhibitor of IGF-1R, and the closely related insulin receptor. Approximately one third of these cell lines were sensitive to OSI-906, with an IC50 < 1 μmol/L. Cell line expression of IGF-1R, IR, IGF-1, IGF-2, IGFBP3, and IGFBP6 did not correlate with sensitivity to OSI-906. Interestingly, OSI-906 sensitive lines expressed significantly lower levels of baseline phospho-ERK relative to resistant lines (P = 0.006). OSI-906 treatment resulted in dose-dependent inhibition of phospho-IGF-1R and phospho-Akt in both sensitive and resistant cell lines, but induced apoptosis and cell-cycle arrest only in sensitive lines. We tested the in vivo efficacy of OSI-906 using an NCI-H187 xenograft model and two SCLC patient xenografts in mice. OSI-906 treatment resulted in 50% tumor growth inhibition in NCI-H187 and 30% inhibition in the primary patient xenograft models compared with mock-treated animals. Taken together our data support IGF-1R inhibition as a viable treatment strategy for a defined subset of SCLC and suggest that low pretreatment levels of phospho-ERK may be indicative of sensitivity to this therapeutic approach.
Collapse
Affiliation(s)
- Rebekah L Zinn
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at the Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Cao H, Wang G, Meng L, Shen H, Feng Z, Liu Q, Du J. Association between circulating levels of IGF-1 and IGFBP-3 and lung cancer risk: a meta-analysis. PLoS One 2012. [PMID: 23185474 PMCID: PMC3501472 DOI: 10.1371/journal.pone.0049884] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background The insulin-like growth factor (IGF) system was documented to play a predominant role in neoplasia. As lung cancer is one of the most malignant cancers, we conducted a meta-analysis in order to investigate the strength of association between circulating IGF-1 and IGFBP-3 levels and lung cancer. Methodology/Principal Findings A systematic literature search was conducted to identify all prospective case-control studies and case-control studies on circulating IGFs and IGFBPs levels. Six nested case-control studies (1 043 case subjects and 11 472 control participants) and eight case-control studies (401 case subjects and 343 control participants) were included in this meta-analysis. Pooled measure was calculated as the inverse variance-weighted mean of the natural logarithm of multivariate adjusted OR with 95% CIs for highest vs. lowest levels to assess the association of circulating IGF-1 and IGFBP-3 concentrations and lung cancer. Standard mean difference (SMD) was also calculated to indicate the difference of the circulating IGF-1 and IGFBP-3 concentrations between the lung cancer case group and the control group. Of the nested case-control studies, ORs for the highest vs. lowest levels of IGF-1 and IGFBP-3 were 1.047 (95% CI: [0.802,1.367], P = 0.736) and 0.960 (95%CI: [0.591,1.559], P = 0.868) respectively; and SMDs were −0.079 (95%CI:[ −0.169, 0.011], P = 0.086) and −0.097 (95%CI:[ −0.264,0.071], P = 0.258) for IGF-1 and IGFBP-3 respectively. As to the case-control studies, SMDs were 0.568 (95%CI:[ −0.035, 1.171], P = 0.065) and −0.780 (95%CI:[ −1.358, −0.201], P = 0.008) for IGF-1 and IGFBP-3 respectively. Conclusions/Significance Inverse association was shown between IGFBP-3 and lung cancer in the case-control studies,and the circulating level of IGFBP-3 underwent a decline during tumorogenesis and development of lung cancer, which suggested IGFBP-3 a promising candidate for the biomarker of lung cancer.
Collapse
Affiliation(s)
- Hongxin Cao
- Institute of Oncology, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Guanghui Wang
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Long Meng
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Hongchang Shen
- Institute of Oncology, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Zhen Feng
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Qi Liu
- Institute of Oncology, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Jiajun Du
- Institute of Oncology, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
- Department of Thoracic Surgery, Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
- * E-mail:
| |
Collapse
|
20
|
Mazzoccoli G, Sothern RB, Pazienza V, Piepoli A, Muscarella LA, Giuliani F, Tarquini R. Circadian Aspects of Growth Hormone–Insulin-Like Growth Factor Axis Function in Patients With Lung Cancer. Clin Lung Cancer 2012; 13:68-74. [DOI: 10.1016/j.cllc.2011.03.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Accepted: 03/29/2011] [Indexed: 10/18/2022]
|
21
|
Chen P, Wang SJ, Wang HB, Ren P, Wang XQ, Liu WG, Gu WL, Li DQ, Zhang TG, Zhou CJ. The distribution of IGF2 and IMP3 in osteosarcoma and its relationship with angiogenesis. J Mol Histol 2011; 43:63-70. [DOI: 10.1007/s10735-011-9370-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 10/16/2011] [Indexed: 01/16/2023]
|
22
|
Increased insulin-like growth factor 1 receptor protein expression and gene copy number in small cell lung cancer. J Thorac Oncol 2011; 5:1905-11. [PMID: 21124078 DOI: 10.1097/jto.0b013e3181f38f57] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE Identification of new therapies in small cell lung cancer (SCLC) is urgently needed. Insulin-like growth factor 1 receptor (IGF1R) is a tyrosine kinase receptor implicated in the pathogenesis of several malignancies and is potentially an attractive target for anticancer treatment. Knowledge about IGF1R protein expression, gene copy number, and the prognostic relevance of these features in SCLC is limited. METHODS We analyzed IGF1R protein expression and gene copy number in primary tumors from 90 patients with SCLC (67 men and 23 women) who underwent pulmonary resection. IGF1R expression assessed by immunohistochemistry with H scores from 0 to 400 was evaluable in 84 patients and IGF1R gene copy number assessed by silver in situ hybridization technique in 81 patients. RESULTS Median H score for IGF1R protein expression was 88 (range, 0-400), and the proportion of positive immunostaining using cutoff H score of 10 was 74%. Increased IGF1R gene copy number (an average of four or more copies per cell) was found in 15 cases (18.5%), five of whom (6.2%) showed gene amplification. There was a significant correlation between protein expression and gene copy number (r = 0.49, p < 0.005). IGF1R expression and gene copy number did not associate with clinicopathological factors such as patient age, tumor size, lymph node involvement, stage, and survival. CONCLUSIONS SCLC is characterized by frequent high-IGF1R protein expression, increased gene copy number, and occasional occurrence of true gene amplification. These features may have important implications for future anti-IGF1R therapeutic approaches.
Collapse
|
23
|
|
24
|
Siwicky MD, Petrik JJ, Moorehead RA. The function of IGF-IR in NNK-mediated lung tumorigenesis. Lung Cancer 2011; 71:11-8. [DOI: 10.1016/j.lungcan.2010.03.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 02/19/2010] [Accepted: 03/21/2010] [Indexed: 11/24/2022]
|
25
|
Abstract
Small-cell lung carcinomas account for about 15-20% of lung cancer and are characterized by an intrinsic resistance to apoptosis. Increasing evidence suggests that alteration in apoptosis/antiapoptosis balance could lead to fundamental resistance of small-cell lung cancer to chemotherapy and radiation. These molecular alterations include alteration of mitochondrial pathways (BCL2 and BCLXL overexpression, activation of stress protein such as HSP 90 and HSP70, activation of PI3K/AKT/mTOR pathway). Others abnormalities could inhibit activation of extrinsic pathway such as caspase-8 and FAS underexpression as well as C-FLIP overexpression. New therapies targeting some of these abnormalities are under clinical evaluation and predictive factors of response are needed to personalize these therapies.
Collapse
|
26
|
Lee W, Patel JH, Lockhart AC. Novel targets in esophageal and gastric cancer: beyond antiangiogenesis. Expert Opin Investig Drugs 2010; 18:1351-64. [PMID: 19642951 DOI: 10.1517/13543780903179286] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cancers of the stomach, gastroesophageal junction and esophagus are a major cause of cancer-related deaths worldwide. In Western countries, adenocarcinomas of the distal esophagus, gastroesophageal junction and proximal stomach have been increasing in frequency more rapidly than other malignancies. The majority of newly diagnosed patients present with advanced disease and the overall survival remains dismal at approximately 10% at 5 years. Better understanding of tumor biology has led to the development of promising novel therapeutic strategies. There is therefore increasing optimism that some of these approaches will improve the outcomes in these increasingly common cancers. Given the success of antiangiogenesis as a therapeutic strategy in various types of cancer, there are ongoing efforts to investigate the utility of other targeted therapies in the treatment of gastric and esophageal cancers. This review will focus on novel therapeutic targets other than angiogenesis and provide a rationale for the further clinical evaluation of these agents in patients with upper gastrointestinal tract cancers.
Collapse
Affiliation(s)
- Wooin Lee
- College of Pharmacy, University of Kentucky, Department of Pharmaceutical Sciences, 725 Rose Street, Room 444, Lexington, KY 40536-0082, USA .
| | | | | |
Collapse
|
27
|
Camidge DR, Dziadziuszko R, Hirsch FR. The rationale and development of therapeutic insulin-like growth factor axis inhibition for lung and other cancers. Clin Lung Cancer 2010; 10:262-72. [PMID: 19632946 DOI: 10.3816/clc.2009.n.037] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The insulin-like growth factor (IGF) axis involves elements of endocrine, paracrine, and autocrine control. It is centrally involved in normal development and growth. Core signaling is driven through the IGF-1 receptor (IGF-1R) in either homo-multimeric complexes or hetero-multimeric complexes with the insulin receptor (IR). Signaling is affected by a large number of upstream and downstream factors, including the differential expression of various intracellular IR substrates, a range of stimulatory ligands (insulin, IGF-1, and IGF-2), the expression of specific clearance receptors (eg, IGF-2R), and different IGF-binding proteins. Considerable evidence exists to implicate aspects of the IGF axis in the development and maintenance of many different nonneoplastic and neoplastic diseases, including both small-cell lung cancer and non-small-cell lung cancer (NSCLC). A large number of different anticancer strategies directed against the IGF axis are being developed. Monoclonal antibodies directed against the IGF-1R are the furthest advanced clinically. Hyperglycemia appears to be a class effect. To date, the major difference among the antibodies used in clinical trials seems to be their plasma half-lives, leading to a number of different administration regimens being taken forward. Early signals of monotherapy activity have been notably reported in patients with Ewing sarcoma and in several other cancers. Encouraging increases in the NSCLC response rate have already been reported after the addition of an anti-IGF-1R antibody to first-line carboplatin and paclitaxel. Explorations of aspects of ligands, binding proteins, receptors, and receptor substrates are all ongoing to identify potential biomarkers predictive of benefit from IGF axis intervention.
Collapse
Affiliation(s)
- D Ross Camidge
- Developmental Therapeutics Program, University of Colorado Cancer Center, Denver Thoracic Oncology Program, University of Colorado Cancer Center, Denver, Colorado 80045, USA.
| | | | | |
Collapse
|
28
|
Findeis-Hosey JJ, Yang Q, Spaulding BO, Wang HL, Xu H. IMP3 expression is correlated with histologic grade of lung adenocarcinoma. Hum Pathol 2009; 41:477-84. [PMID: 20004948 DOI: 10.1016/j.humpath.2009.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 09/30/2009] [Accepted: 10/07/2009] [Indexed: 12/15/2022]
Abstract
Insulin-like growth factor II mRNA binding protein 3 is an oncofetal protein that is expressed in multiple malignancies. This study aimed to determine the correlation of insulin-like growth factor II mRNA binding protein 3 expression with histologic grade of lung adenocarcinoma. Eighty-nine cases, including 11 atypical adenomatous hyperplasias, 10 pure bronchioloalveolar carcinomas, 36 well-differentiated adenocarcinomas and 41 moderately or poorly differentiated adenocarcinomas, were immunohistochemically studied using a monoclonal antibody against insulin-like growth factor II mRNA binding protein 3. Twenty-nine (70.7%) of 41 moderately to poorly differentiated adenocarcinomas were positive for insulin-like growth factor II mRNA binding protein 3, with 26 (89.7%) tumors demonstrating either a strong staining or staining in greater than 30% of tumor cells. Four (40.0%) of 10 bronchioloalveolar carcinomas and 13 (36.1%) of 36 well-differentiated adenocarcinomas exhibited insulin-like growth factor II mRNA binding protein 3 positivity with a variable degree and percentage of tumor cells staining. When bronchioloalveolar carcinomas were present in a pure form or as a component of adenocarcinomas, positive insulin-like growth factor II mRNA binding protein 3 staining was always patchy, with less than 20% of tumor cells stained. Overall, the frequency of positive insulin-like growth factor II mRNA binding protein 3 staining was lower in bronchioloalveolar carcinomas and well-differentiated adenocarcinomas compared to moderately/poorly differentiated adenocarcinomas (P < .01). No insulin-like growth factor II mRNA binding protein 3 signal was detected in any case of atypical adenomatous hyperplasia. These findings show that insulin-like growth factor II mRNA binding protein 3 is strongly and diffusely expressed in a large proportion of moderately/poorly differentiated lung adenocarcinomas, in particular in the solid component of mixed subtype adenocarcinomas, less frequently expressed in well-differentiated adenocarcinomas and bronchioloalveolar carcinomas, and negative in atypical adenomatous hyperplasias. The higher frequency of expression in moderately/poorly differentiated adenocarcinomas suggests that insulin-like growth factor II mRNA binding protein 3 expression may be associated with an aggressive biological behavior.
Collapse
Affiliation(s)
- Jennifer J Findeis-Hosey
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
29
|
Kim WY, Jin Q, Oh SH, Kim ES, Yang YJ, Lee DH, Feng L, Behrens C, Prudkin L, Miller YE, Lee JJ, Lippman SM, Hong WK, Wistuba II, Lee HY. Elevated epithelial insulin-like growth factor expression is a risk factor for lung cancer development. Cancer Res 2009; 69:7439-48. [PMID: 19738076 DOI: 10.1158/0008-5472.can-08-3792] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Insulin-like growth factor (IGF)-I receptor (IGF-IR) signaling has been implicated in several human neoplasms. However, the role of serum levels of IGFs in lung cancer risk is controversial. We assessed the role of tissue-derived IGFs in lung carcinogenesis. We found that IGF-I and IGF-II levels in bronchial tissue specimens containing high-grade dysplasia were significantly higher than in those containing normal epithelium, hyperplasia, and squamous metaplasia. Derivatives of human bronchial epithelial cell lines with activation mutation in KRAS(V12) or loss of p53 overexpressed IGF-I and IGF-II. The transformed characteristics of these cells were significantly suppressed by inactivation of IGF-IR or inhibition of IGF-I or IGF-II expression but enhanced by overexpression of IGF-IR or exposure to the tobacco carcinogens (TC) 4-(methylnitrosamino)-I-(3-pyridyl)-1-butanone and benzo(a)pyrene. We further determined the role of IGF-IR signaling in lung tumorigenesis by determining the antitumor activities of the selective IGF-IR tyrosine kinase inhibitor cis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo [1,5-a]pyrazin-8-ylamine using an in vitro progressive cell system and an in vivo mouse model with a lung-specific IGF-I transgene after exposure to TCs, including 4-(methylnitrosamino)-I-(3-pyridyl)-1-butanone plus benzo(a)pyrene. Our results show that airway epithelial cells produce IGFs in an autocrine or paracrine manner, and these IGFs act jointly with TCs to enhance lung carcinogenesis. Furthermore, the use of selective IGF-IR inhibitors may be a rational approach to controlling lung cancer.
Collapse
Affiliation(s)
- Woo-Young Kim
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Linnerth NM, Siwicky MD, Campbell CI, Watson KLM, Petrik JJ, Whitsett JA, Moorehead RA. Type I insulin-like growth factor receptor induces pulmonary tumorigenesis. Neoplasia 2009; 11:672-82. [PMID: 19568412 PMCID: PMC2697353 DOI: 10.1593/neo.09310] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Revised: 04/13/2009] [Accepted: 04/13/2009] [Indexed: 11/18/2022]
Abstract
Despite the type I insulin-like growth factor receptor (IGF-IR) being highly expressed in more than 80% of human lung tumors, a transgenic model of IGF-IR overexpression in the lung has not been created. We produced two novel transgenic mouse models in which IGF-IR is overexpressed in either lung type II alveolar cells (surfactant protein C [SPC]-IGFIR) or Clara cells (CCSP-IGFIR) in a doxycycline-inducible manner. Overexpression of IGF-IR in either cell type caused multifocal adenomatous alveolar hyperplasia with papillary and solid adenomas. These tumors expressed thyroid transcription factor 1 and Kruppel-like factor 5 in most tumor cells. Similar to our previous work with lung tumors that developed in the mouse mammary tumor virus-IGF-II transgenic mice, the lung tumors that develop in the SPC-IGFIR and CCSP-IGFIR transgenic mice expressed high levels of the cyclic adenosine monophosphate response element binding protein that was localized primarily to the nucleus. Although elevated IGF-IR expression can initiate lung tumor development, tumors can become independent of IGF-IR signaling as IGF-IR down-regulation in established tumors produced tumor regression in some, but not all, of the tumors. These findings implicate IGF-IR as an important initiator of lung tumorigenesis and suggest that the SPC-IGFIR and CCSP-IGFIR transgenic mice can be used to further our understanding of human lung cancer and the role IGF-IR plays in this disease.
Collapse
Affiliation(s)
- Nicolle M Linnerth
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
31
|
Kono SA, Marshall ME, Ware KE, Heasley LE. The fibroblast growth factor receptor signaling pathway as a mediator of intrinsic resistance to EGFR-specific tyrosine kinase inhibitors in non-small cell lung cancer. Drug Resist Updat 2009; 12:95-102. [PMID: 19501013 DOI: 10.1016/j.drup.2009.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2009] [Revised: 05/09/2009] [Accepted: 05/11/2009] [Indexed: 01/25/2023]
Abstract
The EGFR has been targeted through the development of selective tyrosine kinase inhibitors (TKIs) that have proven effective in a subset of non-small cell lung cancer (NSCLC) patients, many bearing gain-of-function EGFR mutations or egfr gene amplification. However, the majority ( approximately 80-90%) of NSCLC patients do not respond to EGFR-specific TKIs and a high rate of acquired resistance to these therapeutics is observed in those that do respond. Thus, EGFR-specific TKIs will not, as single agents, make a high impact on overall lung cancer survival. A number of studies support the activities of other receptor tyrosine kinase pathways including cMet, IGF-1R and FGFRs as mechanisms for both intrinsic and acquired resistance to EGFR TKIs. While the role of cMet and IGF-1R signaling systems as mechanisms of resistance to EGFR TKIs has been widely reviewed in recent years, the potential role of FGFR-dependent signaling as a mechanism for EGFR TKI resistance has more recently emerged and will be highlighted herein. Due to the high degree of homology of FGFRs with VEGFRs and PDGFRs, FGFR-active TKIs already exist via development of VEGFR-targeted TKIs as angiogenesis inhibitors. Thus, these agents could be rapidly advanced into clinical investigations as FGFR inhibitors, either alone or in combination with TKIs selective for EGFR, cMet or IGF-1R as a means to expand the spectrum of NSCLC patients that can be effectively targeted with TKI-directed therapies.
Collapse
Affiliation(s)
- Scott A Kono
- Department of Medicine, University of Colorado at Denver Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
32
|
Nagasaki S, Miki Y, Akahira JI, Suzuki T, Sasano H. 17beta-hydroxysteroid dehydrogenases in human breast cancer. Ann N Y Acad Sci 2009; 1155:25-32. [PMID: 19250189 DOI: 10.1111/j.1749-6632.2008.03682.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Estrogen plays a pivotal role in development and progression of human breast carcinoma. Before menopause the main source of estrogen in women is circulating estrogen secreted from the ovary, but following menopause the source changes to the hormone that is converted from circulating adrenal androgens in peripheral tissues. Therefore, adrenal androgens have to be converted to estrogen to stimulate breast carcinoma cells. In these steps, several enzymes such as aromatase, steroid sulfatase, and 17beta-hydroxysteroid dehydrogenases (17beta-HSDs) are involved in the production of estrogens. The reaction related to 17beta-HSDs activity is one of the last steps of estradiol biosynthesis, and 14 isozymes of 17beta-HSD have been identified at this juncture. The balance of the relative expression levels of 17beta-HSD isozymes in human breast carcinomas is thought to play a pivotal role in supply of estradiol to estrogen receptor positive carcinoma cells. Understanding the character of 17beta-HSD isozymes in human breast carcinoma thus provides important information on the mechanisms of biosynthesis of estradiol in breast carcinoma and for development of a therapeutic agent targeted for inhibition of local estradiol synthesis in breast carcinoma cells. In the present review we summarize the roles played by 17beta-HSDs in human breast carcinoma to obtain a better understanding of the properties of 17beta-HSDs in human breast carcinoma.
Collapse
Affiliation(s)
- Shuji Nagasaki
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | |
Collapse
|
33
|
Vitale G, van Koetsveld PM, de Herder WW, van der Wansem K, Janssen JAMJL, Colao A, Lombardi G, Lamberts SWJ, Hofland LJ. Effects of type I interferons on IGF-mediated autocrine/paracrine growth of human neuroendocrine tumor cells. Am J Physiol Endocrinol Metab 2009; 296:E559-66. [PMID: 19141687 DOI: 10.1152/ajpendo.90770.2008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We recently demonstrated that interferon (IFN)-beta has a more potent antitumor activity than IFN-alpha in BON cells, a neuroendocrine tumor (NET) cell line. The present study showed the role of type I IFNs in the modulation of the insulin-like growth factor (IGF) system in NETs. BON cells expressed IGF-I, IGF-II, IGF-I receptor, and insulin receptor mRNA. In addition, IGF-I and IGF-II stimulated the proliferation of BON cells and induced an inhibition of DNA fragmentation (apoptosis). As evaluated by quantitative RT-PCR, treatment with IFN-alpha (100 IU/ml) or IFN-beta (100 IU/ml) inhibited the expression of IGF-II mRNA (-42% and -65%, respectively, both P < 0.001), whereas IGF-I receptor mRNA was significantly upregulated by IFN-alpha (+28%, P < 0.001) and downregulated by IFN-beta (-47%, P < 0.001). Immunoreactive IGF-II concentration decreased in the conditioned medium during IFN-alpha (-16%, P < 0.05) and IFN-beta (-69%, P < 0.001) treatment. Additionally, IGF-I receptor bioactivity was reduced (-54%) after IFN-beta treatment. Scatchard analysis of (125)I-labeled IGF-I binding to cell membrane of BON cells revealed a dramatic suppression of maximum binding capacity only in the presence of IFN-beta. Finally, the proapoptotic activity of IFN-beta was partially counteracted by the coadministration of IGF-I and IGF-II (both at 50 nM). In conclusion, these data demonstrate that the IGF system has an important role in autocrine/paracrine growth of BON cells. The more potent antitumor activity of IFN-beta compared with IFN-alpha could be explained by several effects on this system: 1) both IFNs inhibit the transcription of IGF-II, but the suppression is significantly higher after IFN-beta than IFN-alpha and 2) only IFN-beta inhibits the expression of IGF-I receptor.
Collapse
Affiliation(s)
- Giovanni Vitale
- Department of Internal Medicine, Erasmus Medical Center, 3015 GE Rotterdam, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Hamatake M, Yoshino I, Tomiyasu M, Miura N, Okazaki H, Ohba T, Takenaka T, Maehara Y. Intratumoral expression of macrophage migration inhibitory factor is correlated with serum C-reactive protein and interleukin-6 in patients with non-small cell lung cancer. Surg Today 2008; 38:921-5. [PMID: 18820867 DOI: 10.1007/s00595-008-3765-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2007] [Accepted: 01/20/2008] [Indexed: 11/24/2022]
Abstract
PURPOSE To investigate if macrophage migration inhibitory factor (MIF) affects systemic inflammatory response in patients with non-small cell lung cancer (NSCLC), we studied the relationship between intratumoral MIF expression and serum levels of C-reactive protein (CRP) and interleukin-6 (IL-6). METHODS mRNAs for MIF and beta-actin, and 18S ribosomal RNA derived from tumor-bearing and nontumor-bearing tissues were quantified by a quantitative reverse transcription-polymerase chain reaction in 35 patients with NSCLC. Spearman's test was used to examine the correlation between the quantities of RNAs and the preoperative serum levels of CRP and IL-6 in the corresponding patients. RESULTS The intratumoral level of MIF mRNA was significantly correlated with the serum levels of CRP (rho = 0.542, P = 0.0016) and IL-6 (rho = 0.532, P = 0.0173). The level of MIF mRNA in the non-tumor-bearing lung also showed a positive, but relatively weak correlation with the serum CRP level (rho = 0.418, P = 0.0148). CONCLUSION Macrophage migration inhibitory factor expression in the tumor site may affect the systemic inflammatory response in patients with NSCLC.
Collapse
Affiliation(s)
- Motoharu Hamatake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Wang HJ, Tashiro SI, Onodera S, Ikejima T. Inhibition of insulin-like growth factor 1 receptor signaling enhanced silibinin-induced activation of death receptor and mitochondrial apoptotic pathways in human breast cancer MCF-7 cells. J Pharmacol Sci 2008; 107:260-9. [PMID: 18635919 DOI: 10.1254/jphs.08054fp] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Silibinin, which had been used as a hepatoprotectant, was shown to have anticancer activity. In this study we investigated the mechanisms of silibinin-induced apoptosis in human breast cancer MCF-7 cells. Expressions of Fas ligand (FasL), Fas-associated death domain protein (FADD), and Bax were significantly up-regulated in silibinin-treated cells, whilst silibinin induced a conspicuous translocation of Bax to mitochondria and release of cytochrome c to the cytosol. Therefore, both the extrinsic Fas death receptor and intrinsic mitochondrial death pathways played essential roles in silibinin-induced apoptosis. It was also found that silibinin markedly decreased protein expression of SIRT1, a mammalian homologue of yeast Sir2, which was proved to have a role in sequestering Bax away from mitochondria. Insulin-like growth factor 1 receptor (IGF-1R), a receptor tyrosine kinase with a crucial role in malignancy development, is expressed in most human primary breast carcinomas. Our results showed that silibinin-induced apoptosis was significantly reinforced by blocking IGF-1R signaling with tyrphostin AG1024, a specific inhibitor of IGF-1R autophosphorylation. Up-regulation of FADD, down-regulation of SIRT1 expression, and activation of the mitochondrial death pathway were apparently enhanced by AG1024 in the silibinin-treated MCF-7 cells.
Collapse
Affiliation(s)
- Hong-Jun Wang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University, Shenyang, China
| | | | | | | |
Collapse
|
36
|
Abstract
Small cell lung carcinoma (SCLC) accounts for approximately 15% of all lung cancer cases. Despite a frequently good response to first-line treatment with chemotherapy and/or radiotherapy, early relapse occurs in the majority of patients and 5-year survival is only about 5%. Therefore, there is a need to develop novel treatments to improve the outcome of patients with SCLC. To fulfil this need, it is critical to gain further understanding on the molecular basis of SCLC and specifically to identify novel therapeutic targets. Clinical trials with molecularly targeted agents have been performed with little success in the past, but recently many promising oncogenic pathways have been discovered and novel targeted therapies are under evaluation. In this review, we summarise the most relevant genetic and signalling pathway alterations reported to date in SCLC and discuss the potential therapeutic implications of such events.
Collapse
|
37
|
Lee CY, Jeon JH, Kim HJ, Shin DH, Roh TW, Ahn CM, Chang YS. Clinical significance of insulin-like growth factor-1 receptor expression in stage I non-small-cell lung cancer: immunohistochemical analysis. Korean J Intern Med 2008; 23:116-20. [PMID: 18787363 PMCID: PMC2686961 DOI: 10.3904/kjim.2008.23.3.116] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND/AIMS The insulin-like growth factor (IGF) system has been implicated in tumor growth, invasion, and metastasis. However, reports on the IGF-1 receptor (IGF-1R) based on radioimmunoassays are conflicting, and its prognostic implications in non-small-cell lung cancer (NSCLC) are still controversial. METHODS Seventy-one paraffin-embedded tissue sections from stage I NSCLC patients were stained using a mouse monoclonal antibody against human IGF-1R. RESULTS The intensity and frequency of IGF-1R expression on the membrane and cytoplasm of cancer cells was evaluated and scored using a semiquantitative system. IGF-1R expression was detected in nine of 71 (12.7%) cases. No significant relationship was found between clinical/histopathological parameters and IGF-1R expression. None of the patients whose tumor expressed IGF-1R had experienced distant metastasis or cancer-related death, although the difference did not reach statistical significance. CONCLUSIONS We conclude that IGF-1R expression may not be a major prognostic factor for stage I NSCLC.
Collapse
Affiliation(s)
- Chang Youl Lee
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Korea
| | - Jeong Hee Jeon
- Human Barrier Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung Jung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Dong Hwan Shin
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Tae Woong Roh
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Chul Min Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Soo Chang
- Human Barrier Research Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Rowinsky EK, Youssoufian H, Tonra JR, Solomon P, Burtrum D, Ludwig DL. IMC-A12, a human IgG1 monoclonal antibody to the insulin-like growth factor I receptor. Clin Cancer Res 2007; 13:5549s-5555s. [PMID: 17875788 DOI: 10.1158/1078-0432.ccr-07-1109] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Targeted monoclonal antibody therapy is an important strategy in cancer therapeutics. Among the most promising characteristics of therapeutic targets are those that modulate the growth and survival of malignant neoplasms and their sensitivity to anticancer therapies. The insulin-like growth factor-I receptor (IGF-IR) is overexpressed in many types of solid and hematopoietic malignancies, and has been implicated as a principal cause of heightened proliferative and survival signaling. IGF-IR has also been shown to confer resistance to cytotoxic, hormonal, and targeted therapies, suggesting that therapeutics targeting IGF-IR may be effective against a broad range of malignancies. IMC-A12 (ImClone Systems Incorporated), a fully human monoclonal IgG1 antibody that binds with high affinity to the IGF-IR, inhibits ligand-dependent receptor activation and downstream signaling. IMC-A12 also mediates robust internalization and degradation of the IGF-IR. In human tumor xenograft models, IGF-IR blockade by IMC-A12 results in rapid and profound growth inhibition of cancers of the breast, lung, colon, and pancreas, and many other neoplasms. Although promising single-agent activity has been observed, the most impressive effects of targeting the IGF-IR with IMC-A12 have been noted when this agent was combined with cytotoxic agents or other targeted therapeutics. The results with IMC-A12 to date suggest that it may be an effective therapeutic in a diverse array of oncologic indications.
Collapse
Affiliation(s)
- Eric K Rowinsky
- Department of Clinical Research and Regulatory Affairs, ImClone Systems Incorporated, Branchburg, New Jersey 08876, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Surinya KH, Forbes BE, Occhiodoro F, Booker GW, Francis GL, Siddle K, Wallace JC, Cosgrove LJ. An investigation of the ligand binding properties and negative cooperativity of soluble insulin-like growth factor receptors. J Biol Chem 2007; 283:5355-63. [PMID: 18056713 DOI: 10.1074/jbc.m707054200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To investigate the interaction of the insulin-like growth factor (IGF) ligands with the insulin-like growth factor type 1 receptor (IGF-1R), we have generated two soluble variants of the IGF-1R. We have recombinantly expressed the ectodomain of IGF-1R or fused this domain to the constant domain from the Fc fragment of mouse immunoglobulin. The ligand binding properties of these soluble IGF-1Rs for IGF-I and IGF-II were investigated using conventional ligand competition assays and BIAcore biosensor technology. In ligand competition assays, the soluble IGF-1Rs both bound IGF-I with similar affinities and a 5-fold lower affinity than that seen for the wild type receptor. In addition, both soluble receptors bound IGF-II with similar affinities to the wild type receptor. BIAcore analyses showed that both soluble IGF-1Rs exhibited similar ligand-specific association and dissociation rates for IGF-I and for IGF-II. The soluble IGF-1R proteins both exhibited negative cooperativity for IGF-I, IGF-II, and the 24-60 antibody, which binds to the IGF-1R cysteine-rich domain. We conclude that the addition of the self-associating Fc domain to the IGF-1R ectodomain does not affect ligand binding affinity, which is in contrast to the soluble ectodomain of the IR. This study highlights some significant differences in ligand binding modes between the IGF-1R and the insulin receptor, which may ultimately contribute to the different biological activities conferred by the two receptors.
Collapse
Affiliation(s)
- Katharina Helen Surinya
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Ji QS, Mulvihill MJ, Rosenfeld-Franklin M, Cooke A, Feng L, Mak G, O'Connor M, Yao Y, Pirritt C, Buck E, Eyzaguirre A, Arnold LD, Gibson NW, Pachter JA. A novel, potent, and selective insulin-like growth factor-I receptor kinase inhibitor blocks insulin-like growth factor-I receptor signaling in vitro and inhibits insulin-like growth factor-I receptor dependent tumor growth in vivo. Mol Cancer Ther 2007; 6:2158-67. [PMID: 17671083 DOI: 10.1158/1535-7163.mct-07-0070] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin-like growth factor-I receptor (IGF-IR) and its ligands, IGF-I and IGF-II, are up-regulated in a variety of human cancers. In tumors, such as colorectal, non-small cell lung, ovarian, and pediatric cancers, which may drive their own growth and survival through autocrine IGF-II expression, the role of IGF-IR is especially critical. Here, we present a novel small-molecule IGF-IR kinase inhibitor, cis-3-[3-(4-methyl-piperazin-l-yl)-cyclobutyl]-1-(2-phenyl-quinolin-7-yl)-imidazo[1,5-a]pyrazin-8-ylamine (PQIP), which displayed a cellular IC(50) of 19 nmol/L for inhibition of ligand-dependent autophosphorylation of human IGF-IR with 14-fold cellular selectivity relative to the human insulin receptor. PQIP showed minimal activity against a panel of 32 other protein kinases. It also abolished the ligand-induced activation of downstream phosphorylated AKT and phosphorylated extracellular signal-regulated kinase 1/2 in both IGF-IR transfectant cells and a GEO human colorectal cancer cell line. Analysis of GEO cells revealed a significant level of both phosphorylated IGF-IR and IGF-II expression. Furthermore, inactivation of IGF-II in conditioned GEO culture medium by a neutralizing antibody diminished IGF-IR activation, indicating the presence of a functional IGF-II/IGF-IR autocrine loop in GEO cells. Once daily oral dosing of PQIP induced robust antitumor efficacy in GEO xenografts. The antitumor efficacy correlated with the degree and duration of inhibition of tumor IGF-IR phosphorylation in vivo by this compound. Moreover, when mice were treated for 3 days with a dose of PQIP that maximally inhibited tumor growth, only minor changes in blood glucose were observed. Thus, PQIP represents a potent and selective IGF-IR kinase inhibitor that is especially efficacious in an IGF-II-driven human tumor model.
Collapse
Affiliation(s)
- Qun-sheng Ji
- Oncology, OSI Pharmaceuticals, Inc., New York, New York, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhu CQ, Popova SN, Brown ERS, Barsyte-Lovejoy D, Navab R, Shih W, Li M, Lu M, Jurisica I, Penn LZ, Gullberg D, Tsao MS. Integrin alpha 11 regulates IGF2 expression in fibroblasts to enhance tumorigenicity of human non-small-cell lung cancer cells. Proc Natl Acad Sci U S A 2007; 104:11754-9. [PMID: 17600088 PMCID: PMC1913903 DOI: 10.1073/pnas.0703040104] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Integrin alpha11 (ITGA11/alpha11) is localized to stromal fibroblasts and commonly overexpressed in non-small-cell lung carcinoma (NSCLC). We hypothesized that stromal alpha11 could be important for the tumorigenicity of NSCLC cells. SV40 immortalized mouse embryonic fibroblasts established from wild-type (WT) and Itga11-deficient [knockout (KO)] mice were tested for their tumorigenicity in immune-deficient mice when implanted alone or coimplanted with the A549 human lung adenocarcinoma cells. A549 coimplanted with the fibroblasts showed a markedly enhanced tumor growth rate compared with A549, WT, or KO, which alone formed only small tumors. Importantly, the growth was significantly greater for A549+WT compared with A549+KO tumors. Reexpression of human alpha11 cDNA in KO cells rescued a tumor growth rate to that comparable with the A549+WT tumors. These findings were validated in two other NSCLC cell lines, NCI-H460 and NCI-H520. Gene expression profiling indicated that IGF2 mRNA expression level was >200 times lower in A549+KO compared with A549+WT tumors. Stable short-hairpin RNA (shRNA) down-regulation of IGF2 in WT (WT(shIGF2)) fibroblasts resulted in a decreased growth rate of A549+WT(shIGF2), compared with A549+WT tumors. The results indicate that alpha11 is an important stromal factor in NSCLC and propose a paradigm for carcinoma-stromal interaction indirectly through interaction between the matrix collagen and stromal fibroblasts to stimulate cancer cell growth.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Transformed
- Cell Line, Tumor
- Collagen/metabolism
- Extracellular Matrix/genetics
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Fibroblasts/metabolism
- Gene Expression Regulation, Neoplastic/physiology
- Growth Substances/biosynthesis
- Growth Substances/genetics
- Growth Substances/physiology
- Humans
- Insulin-Like Growth Factor II
- Integrin alpha Chains/deficiency
- Integrin alpha Chains/genetics
- Integrin alpha Chains/physiology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Mice
- Mice, Knockout
- Mice, SCID
- Proteins/genetics
- Proteins/metabolism
- Stromal Cells/metabolism
- Stromal Cells/pathology
Collapse
Affiliation(s)
- Chang-Qi Zhu
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Svetlana N. Popova
- Department of Biomedicine, Division of Physiology, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; and
| | - Ewan R. S. Brown
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Dalia Barsyte-Lovejoy
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Roya Navab
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Warren Shih
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Ming Li
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Ming Lu
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
| | - Igor Jurisica
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
- Departments of Medical Biophysics
- Computer Science, and
| | - Linda Z. Penn
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
- Departments of Medical Biophysics
| | - Donald Gullberg
- Department of Biomedicine, Division of Physiology, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway; and
| | - Ming-Sound Tsao
- *Ontario Cancer Institute and Princess Margaret Hospital, University Health Network, Toronto, ON, Canada M5G 2M9
- Departments of Medical Biophysics
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada M5G 2M9
- **To whom correspondence should be addressed.
| |
Collapse
|
42
|
Xu H, Bourne PA, Spaulding BO, Wang HL. High-grade neuroendocrine carcinomas of the lung express K homology domain containing protein overexpressed in cancer but carcinoid tumors do not. Hum Pathol 2007; 38:555-63. [PMID: 17316760 DOI: 10.1016/j.humpath.2006.11.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/10/2006] [Accepted: 11/13/2006] [Indexed: 11/30/2022]
Abstract
K homology domain containing protein overexpressed in cancer (KOC) is a member of the insulin-like growth factor (IGF) messenger RNA-binding protein family and is expressed during embryogenesis and in certain malignancies. KOC, known as L523S and IGF messenger RNA-binding protein 3, was shown to be frequently expressed in high-grade neuroendocrine carcinomas of the lung in our immunohistochemical studies using a monoclonal antibody against human KOC. Specifically, all 10 small cell lung carcinomas (SCLCs) exhibited strong cytoplasmic staining, 9 with diffuse positivity and 1 with focal positivity. Among 14 large cell neuroendocrine carcinomas (LCNECs), 9 exhibited strong and diffuse cytoplasmic staining, and 5 cases showed focal immunoreactivity. In contrast, no KOC was detected in 21 typical and atypical carcinoids, except for one atypical carcinoid with oncocytic cells showing weak cytoplasmic staining. Although SCLCs exhibited a strong and diffuse staining pattern more frequently (90%) than LCNECs (64%), the difference did not reach statistical significance (P = .3408). Interestingly, our immunohistochemical studies demonstrated that IGF-II, reportedly regulated by KOC, was comparably expressed in SCLC, LCNEC, and typical and atypical carcinoids, irrespective of KOC expression status of the tumors. These results support the formulation that KOC may play an important role in the regulation of biologic behavior of high-grade neuroendocrine carcinomas. In addition, detection of KOC expression may be diagnostically useful in distinguishing high-grade neuroendocrine carcinomas from carcinoid tumors. Our findings of equivalent IGF-II expression in KOC-positive SCLC and LCNEC and KOC-negative carcinoid tumors suggest different regulatory mechanisms involved in the control of IGF-II expression in these tumors.
Collapse
Affiliation(s)
- Haodong Xu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | |
Collapse
|
43
|
Fischer B, Marinov M, Arcaro A. Targeting receptor tyrosine kinase signalling in small cell lung cancer (SCLC): what have we learned so far? Cancer Treat Rev 2007; 33:391-406. [PMID: 17368733 DOI: 10.1016/j.ctrv.2007.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Revised: 01/15/2007] [Accepted: 01/23/2007] [Indexed: 11/16/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive form of lung cancer, which represents 13% of all cases and is strongly associated with cigarette smoking. The survival of SCLC patients is dismal and has not greatly improved in the last 20 years, despite advances in chemotherapy regimens and a better understanding of SCLC biology. The development of resistance to chemotherapy and metastasis are commonly recognized as important causes of poor clinical outcome in SCLC. Targeting receptor tyrosine kinase (RTK) signalling represents an attractive approach to develop new drugs for SCLC, in view of the accumulating data demonstrating that polypeptide growth factors play a key role in driving SCLC cell proliferation, chemoresistance and metastasis. The insulin-like growth factor-I receptor (IGF-IR), c-Kit, vascular endothelial growth factor receptor (VEGFR) and epidermal growth factor receptor (EGFR) have been identified as potential drug targets in SCLC. Moreover, downstream signalling mediators of RTKs, such as phosphoinositide 3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) may also represent attractive candidate molecules for anti-cancer therapies in SCLC. Here we will review the available data concerning results with RTK inhibitors in SCLC and the clinical trials undertaken to investigate the potential of these compounds as anti-tumour agents in SCLC.
Collapse
Affiliation(s)
- Barbara Fischer
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland.
| | | | | |
Collapse
|
44
|
Allen GW, Saba C, Armstrong EA, Huang SM, Benavente S, Ludwig DL, Hicklin DJ, Harari PM. Insulin-like growth factor-I receptor signaling blockade combined with radiation. Cancer Res 2007; 67:1155-62. [PMID: 17283150 DOI: 10.1158/0008-5472.can-06-2000] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Signaling through the insulin-like growth factor-I receptor (IGF-IR) is implicated in cellular proliferation, apoptosis, carcinogenesis, metastasis, and resistance to cytotoxic cancer therapies. Targeted disruption of IGF-IR signaling combined with cytotoxic therapy may therefore yield improved anticancer efficacy over conventional treatments alone. In this study, a fully human anti-IGF-IR monoclonal antibody A12 (ImClone Systems, Inc., New York, NY) is examined as an adjunct to radiation therapy. IGF-IR expression is shown for a diverse cohort of cell lines, whereas targeted IGF-IR blockade by A12 inhibits IGF-IR phosphorylation and activation of the downstream effectors Akt and mitogen-activated protein kinase. Anchorage-dependent proliferation and xenograft growth is inhibited by A12 in a dose-dependent manner, particularly for non-small cell lung cancer lines. Clonogenic radiation survival of H226 and H460 cells grown under anchorage-dependent conditions is impaired by A12, demonstrating a radiation dose-enhancing effect for IGF-IR blockade. Postradiation anchorage-independent colony formation is inhibited by A12 in A549 and H460 cells. In the H460 xenograft model, combining A12 and radiation significantly enhances antitumor efficacy compared with either modality alone. These effects may be mediated by promotion of radiation-induced, double-stranded DNA damage and apoptosis as observed in cell culture. In summary, these results validate IGF-IR signal transduction blockade as a promising strategy to improve radiation therapy efficacy in human tumors, forming a basis for future clinical trials.
Collapse
Affiliation(s)
- Gregory W Allen
- Department of Human Oncology, School of Medicine and Comprehensive Cancer Center, University of Wisconsin Hospital and Clinics, 600 Highland Avenue, Madison, WI 53792, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Mulvihill MJ, Ji QS, Werner D, Beck P, Cesario C, Cooke A, Cox M, Crew A, Dong H, Feng L, Foreman KW, Mak G, Nigro A, O'Connor M, Saroglou L, Stolz KM, Sujka I, Volk B, Weng Q, Wilkes R. 1,3-Disubstituted-imidazo[1,5-a]pyrazines as insulin-like growth-factor-I receptor (IGF-IR) inhibitors. Bioorg Med Chem Lett 2006; 17:1091-7. [PMID: 17127062 DOI: 10.1016/j.bmcl.2006.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 11/04/2006] [Accepted: 11/07/2006] [Indexed: 01/23/2023]
Abstract
A series of novel 8-amino-1,3-disubstituted-imidazo[1,5-a]pyrazines was designed and synthesized as IGF-IR inhibitors.
Collapse
Affiliation(s)
- Mark J Mulvihill
- (OSI) Oncology, OSI Pharmaceuticals, Inc., 1 Bioscience Park Drive, Farmingdale, NY 11735, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cosaceanu D, Budiu RA, Carapancea M, Castro J, Lewensohn R, Dricu A. Ionizing radiation activates IGF-1R triggering a cytoprotective signaling by interfering with Ku-DNA binding and by modulating Ku86 expression via a p38 kinase-dependent mechanism. Oncogene 2006; 26:2423-34. [PMID: 17043647 DOI: 10.1038/sj.onc.1210037] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Ionizing radiation exposure results in the activation of several tyrosine kinase receptors that participate in radiation-induced DNA damage response and radioresistance. We previously showed that insulin-like growth factor 1 receptor (IGF-1R) inhibition enhanced radiosensitivity of non-small-cell lung cancer (NSCLC) cells. In this paper, we demonstrate that in U1810 NSCLC cells gamma-radiation activates IGF-1R within 10 min, with a maximal activation effect 2 h post-irradiation. Impairment of IGF-1R tyrosine kinase activity enhances human lung cancer cells radiosensitivity by a mechanism that involves phosphatidylinositol 3-kinase (PI3-K) and p38 kinase. In an active form, IGF-1R binds and activates p38 kinase, promoting receptor signaling. Conversely, inhibition of IGF-1R phosphorylation results in IGF-1R/p38 complex disruption and p38 kinase inactivation. We have also demonstrated that in insulin-like growth factor-1-stimulated cells, Ku-DNA-binding activation is induced by ionizing radiation within 4 h, reaches a maximum level at 12 h and remains active up to 72 h. Blockade of IGF-1R activity or its downstream signaling through p38 kinase induces a decrease in radiation-mediated Ku-DNA-binding activation and downregulates the level of Ku86, without affecting Ku70 expression in the nucleus of U1810 cells. The IGF-1R signaling via PI3-K does not interfere with the p38 signaling, the Ku-DNA-binding activity or the level of Ku86. Our present study demonstrates for the first time that ionizing radiation activates IGF-1R. Inhibition of IGF-1R signaling via p38 kinase induces radiosensitivity by a novel mechanism involving nuclear Ku86.
Collapse
Affiliation(s)
- D Cosaceanu
- Department of Oncology-Pathology, Cancer Center Karolinska and Radiumhemmet Karolinska Institute/Hospital, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
47
|
Han JY, Choi BG, Choi JY, Lee SY, Ju SY. The prognostic significance of pretreatment plasma levels of insulin-like growth factor (IGF)-1, IGF-2, and IGF binding protein-3 in patients with advanced non-small cell lung cancer. Lung Cancer 2006; 54:227-34. [PMID: 16935391 DOI: 10.1016/j.lungcan.2006.07.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/20/2006] [Accepted: 07/24/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE Insulin-like growth factor (IGF) system is related to cell proliferation and tumor growth. We tested whether pretreatment plasma levels of IGF-1, IGF-2, and IGF binding protein (IGFBP)-3 would predict the prognosis in patients with advanced non-small cell lung cancer (NSCLC). METHODS Plasma levels of IGF-1, IGF-2, and IGFBP-3 were measured using enzyme-linked immunoassays from 77 patients with advanced NSCLC enrolled in a phase II study of irinotecan plus cisplatin chemotherapy. RESULTS IGF-2 and IGFBP-3 levels were elevated in female patients, non-squamous cell carcinoma, and never smokers. In a univariate Cox proportional hazards model, higher levels of IGF-1, IGF-2, and IGFBP-3 were predictive of longer progression-free (P=0.001, 0.006, and 0.007, respectively) and overall survival (P=0.025, <0.0001, and 0.001, respectively). Multivariate analysis revealed that IGF-1 and IGFBP-3 are independent factors for progression-free survival (P<0.0001 and P=0.001, respectively). In addition, IGF-1, IGF-2, and IGFBP-3 are independently predictive for overall survival (P=0.004, 0.001, and 0.043, respectively). CONCLUSIONS High plasma levels of IGF-1, IGF-2, and IGFBP-3 were associated with good prognosis in patients with advanced NSCLC. Further validation of these results is needed to determine the prognostic significance of IGF system in advanced NSCLC.
Collapse
Affiliation(s)
- Ji-Youn Han
- Research Institute and Hospital, Lung Cancer Branch, National Cancer Center, 809 Madu-dong, Ilsan-gu, Goyang, Gyeonggi 411-769, Republic of Korea.
| | | | | | | | | |
Collapse
|
48
|
Jones HE, Gee JMW, Barrow D, Tonge D, Holloway B, Nicholson RI. Inhibition of insulin receptor isoform-A signalling restores sensitivity to gefitinib in previously de novo resistant colon cancer cells. Br J Cancer 2006; 95:172-80. [PMID: 16819546 PMCID: PMC2360620 DOI: 10.1038/sj.bjc.6603237] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Resistance to antiepidermal growth factor (EGFR) strategies is an emerging clinical problem. Using human colorectal cancer (CRC) cells, we evaluated the involvement of the insulin receptor isoform-A (InsR-A) in de novo resistance to gefitinib, an EGFR tyrosine kinase inhibitor. Challenging the EGFR positive LoVo cells with gefitinib (1 microM) resulted in a small ( approximately 18%) inhibition of cell growth and although a modest reduction in phospho (p)EGFR Tyr845 was seen, pEGFR at residues -Tyr1068 and -Tyr1173 were unchanged. LoVo cells produced unprocessed pro-IGF-1R protein, substantial levels of IGF-II mRNA and mature InsR protein, consisting mainly of the InsR-A isoform. Insulin and IGF-II promoted cell growth and pEGFR Tyr845, Tyr1068 and Tyr1173 activity and conversely, the insulin-like growth factor-1 receptor (IGF-1R)/InsR inhibitor ABDP (1 muM) inhibited growth and reduced pEGFR activity at all three tyrosine residues. pInsR and pAkt levels were increased after gefitinib treatment. Blocking of pInsR with ABDP enabled gefitinib to markedly reduce pEGFR Tyr845, Tyr1068 and Tyr1173. Short-term gefitinib/ABDP dual treatment was more effective than either agent alone and chronic exposure to this combination resulted in total cell loss after 9 weeks, preventing acquisition of resistance to ABDP. LoVo cells with acquired resistance to ABDP were acutely sensitive to gefitinib. We concluded that InsR-A reduces sensitivity to gefitinib in LoVo CRC cells, thus its co-targeting alongside EGFR can improve the anti-tumour effect of gefitinib.
Collapse
Affiliation(s)
- H E Jones
- Tenovus Centre for Cancer Research, Welsh School of Pharmacy, Cardiff University, Cardiff, UK.
| | | | | | | | | | | |
Collapse
|
49
|
Decreased Immunoreactivity of CD99 Is an Independent Predictor of Regional Lymph Node Metastases in Pulmonary Carcinoid Tumors. J Thorac Oncol 2006. [DOI: 10.1097/01243894-200606000-00015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Robinson SA, Rosenzweig SA. Paradoxical effects of the phage display-derived peptide antagonist IGF-F1-1 on insulin-like growth factor-1 receptor signaling. Biochem Pharmacol 2006; 72:53-61. [PMID: 16684509 DOI: 10.1016/j.bcp.2006.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2006] [Revised: 03/30/2006] [Accepted: 03/30/2006] [Indexed: 10/24/2022]
Abstract
The insulin-like growth factor binding proteins (IGFBPs) represent a unique class of IGF antagonists regulating the bioavailability of the IGFs extracellularly. Accordingly, they represent an important class of proteins for cancer therapeutics and chemoprevention. IGF-F1-1 is a cyclic hexadecapeptide identified by high throughput phage display that binds to the IGFBP-binding domain on IGF-1. It acts as an IGFBP-mimetic, capable of inhibiting IGF-1 binding to the IGFBPs. To further examine the utility of IGF-F1-1 as an IGF-1 antagonist we tested its ability to inhibit IGFBP-2 and IGFBP-3 binding to IGF-1, (125)I-IGF-1 binding to IGF-1Rs and to block IGF-1 induced Akt activation, cell cycle changes and [(3)H]thymidine incorporation in MCF-7 cells. These biological activities were inhibited by treatment with IGFBP-2, wortmannin or the IGF-1R tyrosine kinase inhibitor, NVP-AEW541, but not by IGF-F1-1. Our findings confirm previous studies indicating that IGF-F1-1 is a weak antagonist of IGF-1 binding to the IGFBPs and the IGF-1R and suggest that it does not effectively inhibit downstream events stimulated by IGF-1. We further demonstrated that IGF-F1-1 treatment of MCF-7 cells results in the paradoxical activation of Akt, S-phase transition and [(3)H]thymidine incorporation. These results suggest that IGF-F1-1 is a weak agonist, exhibiting mitogenic actions. IGF-F1-1 may act in conjunction with IGF-1 at the IGF-1R or independently of IGF-1 at the IGF-1R or another receptor.
Collapse
Affiliation(s)
- Stephanie A Robinson
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics and Hollings Cancer Center, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | |
Collapse
|